scholarly journals Dynamic relocalization of hOGG1 during the cell cycle is disrupted in cells harbouring the hOGG1-Cys326 polymorphic variant

2005 ◽  
Vol 33 (6) ◽  
pp. 1813-1824 ◽  
Author(s):  
L. Luna
1994 ◽  
Vol 125 (3) ◽  
pp. 625-638 ◽  
Author(s):  
J Lukas ◽  
H Müller ◽  
J Bartkova ◽  
D Spitkovsky ◽  
A A Kjerulff ◽  
...  

The retinoblastoma gene product (pRB) participates in the regulation of the cell division cycle through complex formation with numerous cellular regulatory proteins including the potentially oncogenic cyclin D1. Extending the current view of the emerging functional interplay between pRB and D-type cyclins, we now report that cyclin D1 expression is positively regulated by pRB. Cyclin D1 mRNA and protein is specifically downregulated in cells expressing SV40 large T antigen, adenovirus E1A, and papillomavirus E7/E6 oncogene products and this effect requires intact RB-binding, CR2 domain of E1A. Exceptionally low expression of cyclin D1 is also seen in genetically RB-deficient cell lines, in which ectopically expressed wild-type pRB results in specific induction of this G1 cyclin. At the functional level, antibody-mediated cyclin D1 knockout experiments demonstrate that the cyclin D1 protein, normally required for G1 progression, is dispensable for passage through the cell cycle in cell lines whose pRB is inactivated through complex formation with T antigen, E1A, or E7 oncoproteins as well as in cells which have suffered loss-of-function mutations of the RB gene. The requirement for cyclin D1 function is not regained upon experimental elevation of cyclin D1 expression in cells with mutant RB, while reintroduction of wild-type RB into RB-deficient cells leads to restoration of the cyclin D1 checkpoint. These results strongly suggest that pRB serves as a major target of cyclin D1 whose cell cycle regulatory function becomes dispensable in cells lacking functional RB. Based on available data including this study, we propose a model for an autoregulatory feedback loop mechanism that regulates both the expression of the cyclin D1 gene and the activity of pRB, thereby contributing to a G1 phase checkpoint control in cycling mammalian cells.


2004 ◽  
Vol 45 (7) ◽  
pp. 1437-1443 ◽  
Author(s):  
Maria R Ricciardi ◽  
Maria T Petrucci ◽  
Chiara Gregorj ◽  
Vincenza Martini ◽  
Anna Levi ◽  
...  

1993 ◽  
Vol 105 (2) ◽  
pp. 519-528
Author(s):  
F. Boschelli ◽  
S.M. Uptain ◽  
J.J. Lightbody

The lethal effects of the expression of the oncogenic protein tyrosine kinase p60v-src in Saccharomyces cerevisiae are associated with a loss of cell cycle control at the G1/S and G2/M checkpoints. Results described here indicate that the ability of v-Src to kill yeast is dependent on the integrity of the SH2 domain, a region of the Src protein involved in recognition of proteins phosphorylated on tyrosine. Catalytically active v-Src proteins with deletions in the SH2 domain have little effect on yeast growth, unlike wild-type v-Src protein, which causes accumulation of large-budded cells, perturbation of spindle microtubules and increased DNA content when expressed. The proteins phosphorylated on tyrosine in cells expressing v-Src differ from those in cells expressing a Src protein with a deletion in the SH2 domain. Also, unlike the wild-type v-Src protein, which drastically increases histone H1-associated Cdc28 kinase activity, c-Src and an altered v-Src protein have no effect on Cdc28 kinase activity. These results indicate that the SH2 domain is functionally important in the disruption of the yeast cell cycle by v-Src.


2016 ◽  
Vol 43 (6Part23) ◽  
pp. 3617-3617
Author(s):  
D Sadetaporn ◽  
D Flint ◽  
C McFadden ◽  
A Asaithamby ◽  
G Sawakuchi

Blood ◽  
2013 ◽  
Vol 122 (21) ◽  
pp. 241-241 ◽  
Author(s):  
Daifeng Jiang ◽  
Ricardo Aguiar

Abstract MicroRNA-155 (miR-155) plays pleiotropic roles in the biology of normal and malignant B cells. MiR-155 knockout (KO) mice have fewer germinal center B cells, while overexpression of this miRNA is associated with aggressive DLBCL. Although several miR-155 targets have been identified, a mechanism that unifies the features of loss and gain of miR-155 function in normal and malignant cells remains to be described. In B cells, TGFβ signals are suppressive indicating that deregulation of this pathway may interfere with the developmental regulation of lymphocytes and contribute to the pathogenesis of B cell malignancies. Earlier, we described the direct targeting of the transcription factor SMAD5 by miR-155, and uncovered the presence of non-canonical signaling model in B cell lymphomas whereby TGFβ1, a cytokine that typically activates SMAD2/3, phosphorylated SMAD5. Herein, we used the miR-155 KO mice and genetically modified DLBCL cell lines to investigate which downstream effectors of TGFβ signals are disrupted by the miR-155/SMAD5 interaction, thus shedding light on the phenotypes associates with miR-155 loss and gain of function. We confirmed the phosphorylation of SMAD5 by TGFβ1 in DLBCL cell lines, and demonstrated for the first time that this non-canonical signal is also present in untransformed normal mature B cells. We stably expressed miR-155 in the TGFβ1-responsive DLBCL cell lines Ly7, Ly18 and DHL5, and readily detected suppression of SMAD5, but not of other SMADs. TGFβ1 cytostatic activities include up-regulation of p15 and p21, which are primarily found in the context of SMAD2/3 activation. However, we found that stable expression of miR-155, and downregulation of SMAD5, significantly limited TGFβ1-dependent induction of both p15 and p21 in DLBCL. TGFβ1-mediated upregulation of p15 and p21 limits the activity of cyclin/CDK complexes, enriches for hypophosphorylated (active) RB, and promotes cell cycle arrest. We measured the effects of miR-155 in this process, and found that the accumulation of hypophosphorylated RB following TGFβ1 exposure was blunted in miR-155 expressing cells, resulting in an impaired G0/G1 arrest. The impact of miR-155 on TGFβ1 activity was also detectable by directly measuring the phosphorylation levels of RB’s Ser780 residue. Active pRB blocks cell cycle progression at least in part by binding to and inhibiting the E2F family of transcriptional regulators. Thus, we performed co-immunoprecipitation experiments and quantified the levels of RB-bound E2F1. In these assays, following TGFβ1 exposure we found a markedly decreased pRB-E2F1 complex formation in miR-155 expressing cells when compared to their controls. In agreement with these data, DLBCL cell lines expressing miR-155 displayed higher levels of free E2F1. Together, these data suggested the existence of a miR-155-SMAD5-p15/p21 axis that regulates TGFβ1 effects towards RB and E2F in DLBCL. To confirm the specific role of each component in this circuit, we used an RNAi strategy to transiently or stably knockdown (KD) SMAD5, p15 or p21 in our DLBCL models. In control RNAi cells, exposure to TGFβ1 led to decrease in RB phosphorylation, whereas these effects were abrogated upon KD of each of these genes, resulting in accumulation of hyperphosphorylated RB, a phenocopy of miR-155 expression. To define if the interplay between miR-155/SMAD5 and RB was also present in non-malignant cells, we purified mature B lymphocytes from miR-155 WT and KO mice. Examination of four pairs of mice, showed a higher expression of SMAD5 in cells from miR-155 KO than WT mice. In addition, TGFβ1-mediated suppression of phospho-RB was consistently more pronounced in miR-155 KO than in WT B cells, which resulted in a significantly higher G0/G1 arrest in cells lacking this miRNA. Of note, in absence of TGFβ1 there was no significant difference in cell cycle profile of mature B cells from miR-155 WT and KO mice. We concluded that an unrestrained TGFβ activity, secondary to SMAD5 upregulation, may help explain the deficient germinal center B cells formation found in miR-155 KO mice. Together, our findings demonstrate that miR-155 overexpression is a novel model for deregulation of the lymphomagenic RB/E2F axis, and define an unsuspected role for the non-canonical TGFβ1 activation of SMAD5 in the developmental regulation of mature B cells. Disclosures: No relevant conflicts of interest to declare.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e13563-e13563
Author(s):  
Dennis C. DeSimone ◽  
Trung T. Nguyen ◽  
Eugen Brailiou ◽  
John C. Taylor ◽  
Gabriela Cristina Brailoiu ◽  
...  

e13563 Background: Most ovarian cancer patients are treated with platinum-based chemotherapy but eventually relapse with incurable disease. The G protein-coupled estrogen receptor GPER (GPR30) mediates Ca2+ mobilization in response to estrogen and G-1, a synthetic agonist. Large and sustained Ca2+ responses can lead to mitochondrial Ca2+ overload and apoptosis. Hence, we evaluated whether G-1 could induce apoptosis in cisplatin-sensitive A2780 and isogenic cisplatin–resistant CP70 (14-fold resistant), C30 (70-fold resistant) and C200 (157-fold resistant) human ovarian cancer cells. Bcl-2 and Bcl-xL protect mitochondria from Ca2+overload, and were overexpressed in these cisplatin-resistant cells; thus we also examined combining the Bcl-2 family inhibitor navitoclax with G-1. Methods: Cytoplasmic [Ca2+]c and mitochondrial [Ca2+]m were monitored using microscopy and fluorescent Ca2+ probes. Cell cycle, apoptosis and mitochondrial membrane potential (MMP) were assessed by flow cytometry of propidium iodide, Annexin V and DiIC1(5) -stained cells. The intracellular Ca2+ chelator BAPTA was used to block Ca2+mobilization. Results: Expression of the 53kDa GPER but not the 38 kDa isoform progressively increased with increasing cisplatin resistance. G-1 elicited sustained [Ca2+]c rises that correlated with 53 kDa GPER expression, followed by rises in [Ca2+]m. In all cells, 2.5 μM G-1 blocked cell cycle progression at G2/M, inhibited proliferation, and induced apoptosis (A2780 > C30 > CP70 ≥ C200). G-1 induced p53, caspase-3 and PARP cleavage, and MMP loss. BAPTA prevented G-1’s cell cycle and apoptotic effects in cells showing large Ca2+ mobilization responses but did not in cells with small Ca2+responses. Combining navitoclax with G-1 superadditively decreased cell viability and increased apoptosis. Conclusions: G-1 blocked cell cycle progression and induced apoptosis via a Ca2+-dependent pathway in cells expressing high 53 kDa GPER levels, but via a Ca2+-independent pathway in cells with low 53 kDa GPER expression. G-1 also interacted cooperatively with naviticlax. Therefore, G-1 plus navitoclax shows potential for therapeutic use in platinum-sensitive and -resistant ovarian cancer.


Blood ◽  
2003 ◽  
Vol 101 (10) ◽  
pp. 4078-4087 ◽  
Author(s):  
Qun Liu ◽  
Susan Hilsenbeck ◽  
Yair Gazitt

Abstract Arsenic trioxide (ATO) has been shown to induce differentiation and apoptosis in acute promyelocytic leukemia (APL) cells concomitant with down-regulation of the PML-RARα fusion protein, a product of the t(15:17) translocation characteristic of APL leukemic cells. However, ATO is also a potent inducer of apoptosis in a number of other cancer cells lacking the t(15:17) translocation. The exact mechanism of ATO-induced apoptosis in these cells is not yet clear. We tested the effect of ATO on 7 myeloma cell lines with varying p53 status and report that in cells with mutated p53, ATO induced rapid and extensive (more than 90%) apoptosis in a time- and dose-dependent manner concomitant with arrest of cells in G2/M phase of the cell cycle. Myeloma cells with wild-type (wt) p53 were relatively resistant to ATO with maximal apoptosis of about 40% concomitant with partial arrest of cells in G1 and up-regulation of p21. The use of caspase blocking peptides, fluorescence-tagged caspase-specific substrate peptides, and Western immunoblotting confirmed the involvement of primarily caspase-8 and -3 in ATO-induced apoptosis in myeloma cells with mutated p53 and primarily caspase-9 and -3 in cells expressing wt p53. We also observed up-regulation by ATO of R1 and R2 APO2/TRAIL (tumor necrosis factor–related apoptosis-inducing ligand) receptors. Most important, however, we observed a synergy between ATO and APO2/TRAIL in the induction of apoptosis in the partially resistant myeloma cell lines and in myeloma cells freshly isolated from myeloma patients. Our results justify the use of the combination of these 2 drugs in clinical setting in myeloma patients.


Sign in / Sign up

Export Citation Format

Share Document